Identifying pathways modulating sleep duration: from genomics to transcriptomics

Karla V Allebrandt, Maris Teder-Laving, Paola Cusumano, Goar Frishman, Rosa Levandovski, Andreas Ruepp, Maria P L Hidalgo, Rodolfo Costa, Andres Metspalu, Till Roenneberg, Cristiano De Pittà, Karla V Allebrandt, Maris Teder-Laving, Paola Cusumano, Goar Frishman, Rosa Levandovski, Andreas Ruepp, Maria P L Hidalgo, Rodolfo Costa, Andres Metspalu, Till Roenneberg, Cristiano De Pittà

Abstract

Recognizing that insights into the modulation of sleep duration can emerge by exploring the functional relationships among genes, we used this strategy to explore the genome-wide association results for this trait. We detected two major signalling pathways (ion channels and the ERBB signalling family of tyrosine kinases) that could be replicated across independent GWA studies meta-analyses. To investigate the significance of these pathways for sleep modulation, we performed transcriptome analyses of short sleeping flies' heads (knockdown for the ABCC9 gene homolog; dSur). We found significant alterations in gene-expression in the short sleeping knockdowns versus controls flies, which correspond to pathways associated with sleep duration in our human studies. Most notably, the expression of Rho and EGFR (members of the ERBB signalling pathway) genes was down- and up-regulated, respectively, consistently with the established role of these genes for sleep consolidation in Drosophila. Using a disease multifactorial interaction network, we showed that many of the genes of the pathways indicated to be relevant for sleep duration had functional evidence of their involvement with sleep regulation, circadian rhythms, insulin secretion, gluconeogenesis and lipogenesis.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Figure 1
Figure 1
Human gene vs. disease multifactorial interaction network. The network shows interactions between genes from significant pathways (ERBB signaling family of tyrosine kinases and ion channels) identified based on the GWAS datasets (Meta3, green nodes; Meta7, light-blue nodes; overlapping genes between Meta3 and Meta7, orange nodes) and the Drosophila transcriptome GSEA. Only human homologs of the respective Drosophila genes with altered expression in the dSur KD flies in relation to controls (Fig. 2) were included. Relationships between the respective genes and biological processes (diabetes, carbohydrate and lipid metabolism, orange; cardiovascular diseases, beige), protein complexes and phenotypes (abnormal sleep and circadian behaviour, blue) are also shown (decreased activity/expression, grey edges with cross bar; increased activity/expression, green colored arrows; modulated activity/expression, green edges with open diamond). Red edges indicate gene expression for flies pooled every 3 h of the 24 hours period (decreasing expression, cross bar; increased expression in relation to wild-type controls, arrows; ratio RNAi/wt). Protein-protein interactions are displayed as black edges; interlinking genes are shown as grey nodes. A list of interactions with literature references is available in the Supplementary Table S8.
Figure 2
Figure 2
Differentially expressed genes in dSur KD flies vs. controls. Heat map representing a selection of deregulated transcripts homologs to human genes (indicated in brackets) associated with sleep duration in the meta-analysis results used for the GSEA in (a) “Pooled” (Drosophila pooled every 3 h of the 24 hours period) and (b) “Night” (3 h into the night) conditions. A color-coded scale for the normalized expression values is used as follows: yellow and blue represent high and low expression levels in dSur KD with respect to control, respectively. The expression level of each transcript was calculated as the log2 (dSur KD/control), and the complete lists of differentially expressed genes identified by LIMMA software are provided in the Supplementary Table S7.
Figure 3
Figure 3
Signalling by Rho GTPases (upper panel) and Neuronal system pathway (bottom panel). These Reactome pathways show statistically significant enrichment (Fisher Exact test) of DEGs both in “Night” (a, upper panel left, q-value < 0.004; a, bottom panel left, q-value = 0.007) and “Pooled” (b, upper panel right, q-value < 0.001; b, bottom panel right, q-value < 0.001) conditions analysed with Graphite web tool. Coloured nodes represent the differentially expressed genes. The colour of the nodes is proportional to their expression levels represented as log2 (dSur KD/control). Raw p-values were adjusted using Benjamini-Hockberg method (q-value).

References

    1. Allebrandt KV, et al. Chronotype and sleep duration: the influence of season of assessment. Chronobiol. Int. 2014;31:731–40. doi: 10.3109/07420528.2014.901347.
    1. Wulff K, Gatti S, Wettstein JG, Foster RG. Sleep and circadian rhythm disruption in psychiatric and neurodegenerative disease. Nat. Rev. Neurosci. 2010;11:589–599. doi: 10.1038/nrn2868.
    1. Cappuccio FPF, D’Elia L, Strazzullo P, Miller MMA. Sleep Duration and All-Cause Mortality: A Systematic Review and Meta-Analysis of Prospective Studies. Sleep. 2010;33:585–92. doi: 10.1093/sleep/33.5.585.
    1. Laposky AD, Bass J, Kohsaka A, Turek FW. Sleep and circadian rhythms: key components in the regulation of energy metabolism. FEBS Lett. 2008;582:142–151. doi: 10.1016/j.febslet.2007.06.079.
    1. Allebrandt KV, et al. A KATP channel gene effect on sleep duration: from genome-wide association studies to function in Drosophila. Mol. Psychiatry. 2013;18:122–132. doi: 10.1038/mp.2011.142.
    1. Akrouh A, Halcomb SE, Nichols CG, Sala-Rabanal M. Molecular biology of KATP channels and implications for health and disease. IUBMB Life. 2009;61:971–978. doi: 10.1002/iub.246.
    1. Maret S, et al. Homer1a is a core brain molecular correlate of sleep loss. Proc. Natl. Acad. Sci. USA. 2007;104:20090–20095. doi: 10.1073/pnas.0710131104.
    1. Zhang, K., Cui, S., Chang, S., Zhang, L. & Wang, J. i-GSEA4GWAS: A web server for identification of pathways/gene sets associated with traits by applying an improved gene set enrichment analysis to genome-wide association study. Nucleic Acids Res. 38 (2010).
    1. Kryger, M. H. b., Roth, T. d. e. & Dement, W. C. Principles and Practice of Sleep Medicine. Principles and Practice of Sleep Medicine (2005).
    1. Kantermann T, Juda M, Merrow M, Roenneberg T. The Human Circadian Clock’s Seasonal Adjustment Is Disrupted by Daylight Saving Time. Curr. Biol. 2007;17:1996–2000. doi: 10.1016/j.cub.2007.10.025.
    1. Allebrandt KV, et al. CLOCK Gene Variants Associate with Sleep Duration in Two Independent Populations. Biol. Psychiatry. 2010;67:1040–1047. doi: 10.1016/j.biopsych.2009.12.026.
    1. Marchini J, Howie BN, Myers S, McVean G, Donnelly P. A new multipoint method for genome-wide association studies by imputation of genotypes. Nat. Genet. 2007;39:906–13. doi: 10.1038/ng2088.
    1. Johnson RC, et al. Accounting for multiple comparisons in a genome-wide association study (GWAS) BMC Genomics. 2010;11:724. doi: 10.1186/1471-2164-11-724.
    1. Lechner M, et al. CIDeR: multifactorial interaction networks in human diseases. Genome Biol. 2012;13:R62. doi: 10.1186/gb-2012-13-7-r62.
    1. Shaw PJ, et al. Correlates of sleep and waking in Drosophila melanogaster. Science. 2000;287:1834–7. doi: 10.1126/science.287.5459.1834.
    1. Bolstad BM, Irizarry R, Astrand M, Speed TP. A comparison of normalization methods for high density oligonucleotide array data based on variance and bias. Bioinformatics. 2003;19:185–193. doi: 10.1093/bioinformatics/19.2.185.
    1. Saeed A, et al. TM4 microarray software suit. Methods Enzymol. 2006;411:134–193. doi: 10.1016/S0076-6879(06)11009-5.
    1. Gentleman, R., Carey, V., Huber, W., Irizarry, R. & Dudoit, S. Bioinformatics and Computational Biology Solutions Using R and Bioconductor. Statistics for Biology and Health (2005).
    1. Sales G, Calura E, Cavalieri D, Romualdi C. graphite - a Bioconductor package to convert pathway topology to gene network. BMC Bioinformatics. 2012;13:20. doi: 10.1186/1471-2105-13-20.
    1. Vastrik I, et al. Reactome: a knowledge base of biologic pathways and processes. Genome Biol. 2007;8:R39. doi: 10.1186/gb-2007-8-3-r39.
    1. Gottlieb, D. J. et al. Novel loci associated with usual sleep duration: the CHARGE Consortium Genome-Wide Association Study. Mol. Psychiatry 1–8, doi:10.1038/mp.2014.133 (2014).
    1. Cirelli C. The genetic and molecular regulation of sleep: from fruit flies to humans. Nat. Rev. Neurosci. 2009;10:549–60. doi: 10.1038/nrn2683.
    1. Cavaliere, S. & Hodge, J. J. L. Drosophila KCNQ channel displays evolutionarily conserved electrophysiology and pharmacology with mammalian KCNQ channels. PLoS One6 (2011).
    1. Liguori R, et al. Morvan’s syndrome: peripheral and central nervous system and cardiac involvement with antibodies to voltage-gated potassium channels. Brain. 2001;124:2417–2426. doi: 10.1093/brain/124.12.2417.
    1. Deboer T, van Diepen HC, Ferrari MD, Van den Maagdenberg AMJM, Meijer JH. Reduced sleep and low adenosinergic sensitivity in cacna1a R192Q mutant mice. Sleep. 2013;36:127–36. doi: 10.5665/sleep.2316.
    1. Macedo PG, et al. Sleep-disordered breathing in patients with the Brugada syndrome. Am J Cardiol. 2011;107:709–713. doi: 10.1016/j.amjcard.2010.10.046.
    1. Citri A, Yarden Y. EGF-ERBB signalling: towards the systems level. Nat. Rev. Mol. Cell Biol. 2006;7:505–16. doi: 10.1038/nrm1962.
    1. Foltenyi K, Greenspan RJ, Newport JW. Activation of EGFR and ERK by rhomboid signaling regulates the consolidation and maintenance of sleep in Drosophila. Nat. Neurosci. 2007;10:1160–7. doi: 10.1038/nn1957.
    1. Kushikata T, Fang J, Chen Z, Wang Y, Krueger JM. Epidermal growth factor enhances spontaneous sleep in rabbits. Am. J. Physiol. 1998;275:R509–14.
    1. Bang AG, Kintner C. Rhomboid and star facilitate presentation and processing of the Drosophila TGF-?? homolog Spitz. Genes Dev. 2000;14:177–186.
    1. Nakagawa T, et al. Characterization of a human rhomboid homolog, p100hRho/RHBDF1, which interacts with TGF-?? family ligands. Dev. Dyn. 2005;233:1315–1331. doi: 10.1002/dvdy.20450.
    1. Sun H, et al. Label-free cell phenotypic profiling decodes the composition and signaling of an endogenous ATP-sensitive potassium channel. Sci. Rep. 2014;4:4934. doi: 10.1038/srep04934.
    1. Nichols CG, Singh GK, Grange DK. KATP channels and cardiovascular disease: Suddenly a syndrome. Circulation Research. 2013;112:1059–1072. doi: 10.1161/CIRCRESAHA.112.300514.
    1. Benter IF, et al. Early inhibition of EGFR signaling prevents diabetes-induced up-regulation of multiple gene pathways in the mesenteric vasculature. Vascul. Pharmacol. 2009;51:236–245. doi: 10.1016/j.vph.2009.06.008.
    1. Balasubramanian N, Slepak VZ. Light-mediated activation of Rac-1 in photoreceptor outer segments. Curr. Biol. 2003;13:1306–1310. doi: 10.1016/S0960-9822(03)00511-6.
    1. Taheri S, Lin L, Austin D, Young T, Mignot E. Short sleep duration is associated with reduced leptin, elevated ghrelin, and increased body mass index. PLoS Med. 2004;1:e62. doi: 10.1371/journal.pmed.0010062.
    1. Roenneberg T, Allebrandt KV, Merrow M, Vetter C. Social jetlag and obesity. Curr. Biol. 2012;22:939–943. doi: 10.1016/j.cub.2012.03.038.
    1. Thimgan MS, Seugnet L, Turk J, Shaw PJ. Identification of Genes Associated with Resilience/Vulnerability to Sleep Deprivation and Starvation in Drosophila. Sleep. 2015;38:801–814. doi: 10.5665/sleep.4680.
    1. Thimgan MS, Suzuki Y, Seugnet L, Gottschalk L, Shaw PJ. The Perilipin homologue, Lipid storage droplet 2, regulates sleep homeostasis and prevents learning impairments following sleep loss. PLoS Biol. 2010;8:29–30. doi: 10.1371/journal.pbio.1000466.

Source: PubMed

3
Se inscrever